US20140336129A1 - Targeting the EGFR-SGLT1 Interaction for Cancer Therapy - Google Patents

Targeting the EGFR-SGLT1 Interaction for Cancer Therapy Download PDF

Info

Publication number
US20140336129A1
US20140336129A1 US14/273,375 US201414273375A US2014336129A1 US 20140336129 A1 US20140336129 A1 US 20140336129A1 US 201414273375 A US201414273375 A US 201414273375A US 2014336129 A1 US2014336129 A1 US 2014336129A1
Authority
US
United States
Prior art keywords
egfr
seqid
sglt1
cells
sglt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US14/273,375
Other versions
US9334307B2 (en
Inventor
Zhang Weihua
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Houston System
Original Assignee
University of Houston System
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Houston System filed Critical University of Houston System
Priority to US14/273,375 priority Critical patent/US9334307B2/en
Publication of US20140336129A1 publication Critical patent/US20140336129A1/en
Assigned to THE UNIVERSITY OF HOUSTON SYSTEM reassignment THE UNIVERSITY OF HOUSTON SYSTEM ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZHANG, WEIHUA
Priority to US15/139,066 priority patent/US9770482B2/en
Application granted granted Critical
Publication of US9334307B2 publication Critical patent/US9334307B2/en
Assigned to UNITED STATES GOVERNMENT reassignment UNITED STATES GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF HOUSTON
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/179Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids

Definitions

  • Epidermal growth factor receptor is a receptor tyrosine kinase that is over-active/over-expressed in the majority of cancers of epithelial origin (Hynes N E, et al., ERBB receptors and cancer: the complexity of targeted inhibitors, Nature Reviews Cancer, 5(5):341-354 (2005)). Inhibition of the tyrosine kinase activity of EGFR has been the principle strategy of EGFR based cancer therapies. However, targeting EGFR by small molecule inhibitors of receptor tyrosine kinase has not produced satisfactory therapeutic efficacy.
  • the general response rates are between 10-20% across a variety of human malignancies (Weiss J., First line erlotinib for NSCLC patients not selected by EGFR mutation: keep carrying the TORCH or time to let the flame die? Transl. Lung Cancer Res., 1(3):219-223 (2012); Cohen S J, et al., Phase II and pharmacodynamic study of the farnesyltransferase inhibitor R115777 as initial therapy in patients with metastatic pancreatic adenocarcinoma, J. Clinical Oncology, 21(7):1301-1306 (2003); Dancey J E, et al., Targeting epidermal growth factor receptor—are we missing the mark?, Lancet 362(9377):62-64 (2003)).
  • mice with severely compromised EGFR tyrosine kinase activity are completely viable and display only some epithelial defects (Luetteke N C, et al., The mouse waved-2 phenotype results from a point mutation in the EGF receptor tyrosine kinase, Genes & Development, 8(4):399-413 (1994)).
  • both a wild type and a kinase-dead EGFR enhanced the survival of EGFR negative 32D hematopoietic cells (Ewald J A, et al., Ligand- and kinase activity-independent cell survival mediated by the epidermal growth factor receptor expressed in 32D cells, Experimental Cell Research 282(2):121-131 (2003).
  • EGFR participates in the maintenance of basal intracellular glucose level of cancer cells by interacting with and stabilizing the sodium-glucose co-transporter 1 (SGLT1), independent of EGFR tyrosine kinase activity (Weihua Z, et al., Survival of cancer cells is maintained by EGFR independent of its kinase activity, Cancer Cell, 13(5):385-393 (2008)).
  • SGLT1 sodium-glucose co-transporter 1
  • SGLT1 is an active glucose transporter that relies on extracellular sodium concentration to transport glucose into cells independent of glucose concentration (Wright E M, et al., Biology of human sodium glucose transporters, Physiological Reviews, 91(2):733-794 (2011). SGLT1 plays a critical role in glucose absorption and retention in the body (Castaneda-Sceppa C, et al., Sodium-dependent glucose transporter protein as a potential therapeutic target for improving glycemic control in diabetes, Nutrition Reviews, 69(12):720-729 (2011)).
  • One of the hallmarks of cancer is that cancer cells exhibit altered energy metabolism, i.e.
  • cancer cells consume a substantially higher amount of nutrients and energy substrates than their normal counterparts (Hanahan D, et al., Hallmarks of cancer: the next generation, Cell, 144(5):646-674 (2011).
  • This enhanced energy consumption demands a high rate of nutrients uptake, which is achieved by over-expression of plasma membrane transporters (Ganapathy V, et al., Nutrient transporters in cancer: relevance to Warburg hypothesis and beyond, Pharmacology & Therapeutics 121(1):29-40 (2009).
  • SGLT1 is over-expressed in various types of cancers including ovarian carcinoma, oral squamous cell carcinoma, colorectal cancer, pancreatic cancer, and prostate cancer
  • Lai B, et al. Overexpression of SGLT1 is correlated with tumor development and poor prognosis of ovarian carcinoma, Archives of Gynecology and Obstetrics, 285(5):1455-1461 (2012); Hanabata Y, et al., Coexpression of SGLT1 and EGFR is associated with tumor differentiation in oral squamous cell carcinoma, Odontology/the Society of the Nippon Dental University, 100(2):156-163 (2012); Guo G F, et al., Overexpression of SGLT1 and EGFR in colorectal cancer showing a correlation with the prognosis, Medical Oncology 28 Suppl 1:S197-203 (2011); Casneuf V F, et al., Expression of SGLT1, Bcl-2 and p53 in primary pancreatic
  • FIG. 1A illustrates a schematic diagram of different constructs of human EGFR, in accordance with embodiments.
  • FIG. 1B illustrates an immunoprecipitation coupled Western blot analysis of interactions between mutated EGFRs and SGLT1, in accordance with embodiments.
  • FIG. 2A illustrates a Western blot analysis of expression levels of SGTL1 in HEK293 cells co-transfected with the WT-EGFR, the KD-EGFR and the ⁇ Autophos-EGFR, in accordance with embodiments.
  • FIG. 2B illustrates a densitometric quantification of bands in the Western blot of FIG. 2A , in accordance with embodiments.
  • FIG. 2C illustrates a Western blot analysis of the effect of a proteasome inhibitor MG132 on the down-regulation of SGLT1 by ⁇ Autophos-EGFR, in accordance with embodiments.
  • FIG. 2D illustrates a densitometric quantification of bands in the Western blot of FIG. 2C , in accordance with embodiments.
  • FIG. 3A illustrates an immunoprecipitation coupled Western blot analysis of interactions between EGFR-HA and SGLT1-Flag in HEK293 cells treated with EGF or AEE788, in accordance with embodiments.
  • FIG. 3B illustrates immunoprecipitation coupled Western blot analysis of interactions between endogenous EGFR and SGLT1 in PC3 cells treated with EGF or AEE788, in accordance with embodiments.
  • FIG. 4A illustrates co-localization of SGLT1 and EGFR in prostate cancer tissues from a prostate cancer tissue array, in accordance with embodiments.
  • FIG. 4B illustrates a Western blot analysis of expressions of endogenous EGFR and SGLT1 in PC3 and LNCaP cells, in accordance with embodiments.
  • FIG. 4C illustrates an MTT assay showing the effect of inhibition of SGLT1 on the growth inhibitory effect of Gefitinib and Erlotinib on PC3 cells, in accordance with embodiments.
  • FIG. 4D illustrates an MTT assay showing the effect of inhibition of SGLT1 on the growth inhibitory effect of Gefitinib and Erlotinib on LNCaP cells, in accordance with embodiments.
  • FIG. 5A illustrates disruption of EGFR-SGLT1 interaction by small molecules of peptides to destabilize both proteins, in accordance with embodiments.
  • FIG. 5B illustrates that treatment of PC3 cells with an EGFR-SGLT1 disrupting peptide (MTG-01) significantly down-regulates EGFR and SGLT1 proteins, in accordance with embodiments.
  • MVG-01 EGFR-SGLT1 disrupting peptide
  • FIG. 6 illustrates the viability of cancerous PC3, Du145, HCT116, and MDA-MB-231 cells and treatment of non-cancerous HEK293 cells when treated with MTG-01, in accordance with embodiments.
  • a compound destabilizes a binding interaction between an epidermal growth factor receptor (EGFR) and a sodium/glucose co-transporter 1 (SGLT 1).
  • EGFR epidermal growth factor receptor
  • SGLT 1 sodium/glucose co-transporter 1
  • the compound is a Phlorizin-like compound or a peptide derived from the interacting domain of EGFR.
  • a compound that destabilizes the binding interaction between EGFR and SGLT 1 is administered to a subject intratumorally, intravenously, or systemically.
  • the compound may be a Phlorizin-like compound or a peptide derived from the interacting domain of EGFR.
  • the compound may be administered in conjunction with a tyrosine-kinase inhibitor.
  • EGFR epidermal growth factor receptor
  • SGLT1 Sodium/glucose co-transporter 1
  • SGLT1 is an active glucose transporter that is over-expressed in cancers including prostate cancer. It has been found that EGFR interacts with and stabilizes SGLT1 in cancer cells.
  • the autophosphorylation region (978-1210 amino acids) of EGFR is required for its sufficient interaction with SGLT1.
  • This interaction is independent of EGFR's tyrosine kinase activity.
  • the EGFR-SGLT1 interaction is irresponsive to EGFR tyrosine kinase modulators (EGF and tyrosine kinase inhibitors).
  • EGFR and SGLT1 co-localize in prostate cancer tissues.
  • inhibition of SGLT1 by a SGLT1 inhibitor (Phlorizin) sensitizes prostate cancer cells (PC3 and LNCaP) to EGFR inhibitors (Gefitnib and Erlotinib).
  • ESD-01 destabilizes the EGFR-SGLT1 interaction.
  • SGLT1 is a protein involved in EGFR's functions that are irresponsive to EGFR tyrosine kinase inhibitors, and the EGFR-SGLT1 interaction may be a novel target for prostate cancer therapy.
  • FIG. 1A illustrates a schematic diagram of human EGFR constructs that may be used to determine the EGFR protein region required for its interaction with SGLT1.
  • the constructs may include: wild type EGFR (“WT”); kinase dead EGFR (R817M) (“KD”) (SEQID: 012); transmembrane domain deletion (645-670aa) (“ATM”) (SEQID: 013); extracellular domain deletion (1-644aa) (“ ⁇ Extra”) (SEQID: 014); intracellular domain deletion (671-1210aa) (“ ⁇ Intra”) (SEQID: 015); tyrosine kinase domain deletion (670-977aa) (“ ⁇ TK”) (SEQID: 016); or autophosphorylation domain deletion (978-1210aa) (“ ⁇ Autophos”) (SEQID: 017).
  • the flagged SGLT1 and above identified HA tagged EGFRs can be transiently co-transfected into HEK293 cells.
  • SGLT1 can be immunoprecipitated using anti-flag antibodies.
  • Western blot analyses may be performed for HA tagged EGFRs.
  • deletion of the entire intracellular domain or the autophosphorylation domain of EGFR substantially diminishes its interaction with SGLT1.
  • the autophosphorylation domain of EGFR is required for its sufficient interaction with SGLT1.
  • the TM domain may be included into the constructs of truncated EGFRs.
  • the TM containing intracellular domain of EGFR especially the autophosphorylation domain, interacts more strongly with SGLT1 than its extracelluar domain. The discrepancy between this embodiment and the data shown in the previous report is very likely due to the lack of TM domain in the intracellular domain construct used in the previous study.
  • the expression level of SGLT1 co-transfected with the WT-EGFR, the KD-EGFR, and the ⁇ AutoPhos-EGFR into HEK293 cells may be measured.
  • FIG. 2A illustrates a Western blot analysis of expression levels of SGTL1 in HEK293 cells co-transfected with the WT-EGFR, the KD-EGFR and the ⁇ Autophos-EGFR.
  • the same amounts of DNA plasmids of SGLT1 and EGFRs may be used in each group of treatments.
  • control cells can be transfected with the same amount of DNA of the empty vector. Actin may be used as loading control.
  • FIG. 2B illustrates a densitometric quantification of bands in the Western blot of FIG. 2A . Asterisk marks indicate statistical significance between the linked representative groups from triplicate experiments.
  • the level of SGLT1 in the WT-EGFR and the KD-EGFR transfected cells is much higher than that in the control vector or ⁇ AutoPhos-EGFR transfected cells.
  • the autophosphorylation domain of EGFR can maintain the expression level of SGLT1.
  • the level of SGLT1 in the ⁇ AutoPhos-EGFR transfected cells is significantly lower than that of the control cells.
  • the loss of SGLT1 interaction with EGFR can promote down-regulation of SGLT1.
  • FIG. 2C illustrates a Western blot analysis of the effect of a proteasome inhibitor MG132 on the down-regulation of SGLT1 by ⁇ Autophos-EGFR. Actin may be used as a loading control.
  • FIG. 2D illustrates a densitometric quantification of bands in the Western blot of FIG. 2C .
  • Asterisk marks indicate statistical significance between the linked representative groups from triplicate experiments.
  • MG231 can inhibit the down-regulation of SGLT1 in ⁇ AutoPhos-EGFR transfected cells.
  • the proteasome machinery is involved in the loss of interaction with EGFR induced SGLT1 down-regulation. In other words, proteasomes may cause the ultimate degradation of SGLT1 when the EGFR-SLGT1 interaction is compromised.
  • the proteasome machinery may be a potential therapeutic target alone or in combination with the EGFR-SLGT1 interaction.
  • deleting the SGLT1 interacting domain in EGFR promotes the down-regulation of SGTL1 via the proteasome machinery.
  • this disruption of the EGFR-SGLT1 interaction in EGFR positive cancer cells can lead to down-regulation of SGLT1.
  • previous data shows that knocking down SGLT1 by shRNA results in autophagic cell death of prostate cancer cells (Weihua Z, et al., Survival of cancer cells is maintained by EGFR independent of its kinase activity, Cancer Cell 13(5):385-393 (2008).
  • the EGFR-SGLT1 interaction can be a significant target to improve EGFR based therapy for cancer.
  • EGFR-SGLT1 Interaction is Irresponsive to EGFR Tyrosine Kinase Inhibitors
  • the effects of EGFR tyrosine kinase inhibitors on EGFR's interaction with SGLT1 can be determined.
  • WT-EGFR and SGLT1 co-transfected HEK293 cells can be treated with either EGF or an EGFR tyrosine kinase inhibitor, AEE788.
  • SGLT1 may be immunoprecipitated, and the levels of EGFR that were co-immunoprecipitated with SGLT1 can be measured. For example, FIG.
  • neither EGF nor AEE788 significantly affects the EGFR-SGLT1 interaction.
  • the EGFR that co-precipitates with SGLT1 is not phosphorylated.
  • endogenous SGLT1 of PC3 cells treated with EGF or AEE788 may be immunoprecipitated.
  • the phosphorylation status of the EGFR co-precipitated with SGLT1 can be measured.
  • FIG. 3B illustrates an immunoprecipitation coupled Western blot analysis of interactions between endogenous EGFR and SGLT1 in PC3 cells treated with EGF or AEE788.
  • neither EGF nor AEE788 affect the EGFR-SGLT1 interaction.
  • the endogenous SGLT1 interacting EGFR is not phosphorylated.
  • the EGFR-SGLT1 interaction is irresponsive to modulators of EGFR's tyrosine kinase activity.
  • EGFR can be targeted for cancer therapy at its non-kinase functionality, including but not limited to its interaction with SGLT1.
  • the autophosphorylation domain serves as a major docking site for recruitment of adaptor/effector proteins that transactivate downstream signaling (Bazley L A, et al., The epidermal growth factor receptor family, Endocrine-Related Cancer, 12 Suppl 1:S17-27 (2005)).
  • the EGFR-SGLT1 interaction is independent of EGFR activation/inactivation.
  • the autophosphorylation domain of EGFR functions as a protein-protein interacting domain independent of EGFR's tyrosine kinase activity.
  • EGFR possesses pro-survival functions independent of its tyrosine kinase activity.
  • EGFR can exist in two types of statuses—a tyrosine kinase responsive status and a tyrosine kinase irresponsive status.
  • the autophosphorylation domain of the kinase responsive EGFR can be phosphorylated and can recruit effectors to trigger downstream signals.
  • the kinase irresponsive EGFR may constantly interact with proteins regardless of the presence of EGFR ligands, and regardless of activation or inactivation of its tyrosine kinase.
  • SGLT1 may be one of such proteins that bind to and keep EGFRs in their kinase irresponsive status.
  • the non-phosphorylated autophosphorylation domain of EGFR may be used as a tool for identifying therapeutic targets.
  • FIG. 4A illustrates the results of three representative prostate cancer tissues from a prostate cancer tissue array.
  • SGLT1 green
  • EGFR red
  • stromal cells may be positive for SGLT1, but negative for EGFR (e.g., indicated by arrow heads).
  • SGLT1 co-localizes with EGFR in cancer cells, but not stromal cells.
  • the EGFR-SGLT1 interaction may contribute to the pathogenesis of prostate cancer.
  • the co-localization of EGFR with SGLT1 in prostate cancer tissues can indicate that the EGFR-SGLT1 interaction is cancer relevant.
  • EGFR tyrosine kinase inhibitors have not yet shown satisfactory therapeutic effects for prostate cancer (Canil C M, et al., Randomized phase II study of two doses of gefitinib in hormone-refractory prostate cancer: a trial of the National Cancer Institute of Canada-Clinical Trials Group, J.
  • EGFR may contribute to the disease progression of prostate cancer independent of its tyrosine kinase activity.
  • prostate cancer tissues have increased expression of SGLT1, 23 (2) a loss of EGFR protein but not its tyrosine kinase activity sensitized prostate cancer cells to chemotherapeutic agent, 29 and (3) a loss of EGFR induced autophagic cell death was mediated by down-regulation of SGLT1 protein (Blessing A, et al., Sodium/Glucose Co-transporter 1 Expression Increases in Human Diseased Prostate, J. Cancer Sci. Ther. 4(9):306-312 (2012); Xu S, et al., Loss of EGFR induced autophagy sensitizes hormone refractory prostate cancer cells to adriamycin, The Prostate.
  • EGFR can promote prostate cancer progression via stabilizing SGLT1 to sustain the high demand of glucose of late stage cancer cells.
  • This embodiment is not only supported by all the embodiments described herein, but is also supported by past data that over-expression of SGLT1 prevented renal epithelial cells and intestinal epithelial cells from apoptosis (Ikari A, et al., Sodium-dependent glucose transporter reduces peroxynitrite and cell injury caused by cisplatin in renal tubular epithelial cells, Biochimica et Biophysica Acta 1717(2):109-117 (2005); Yu L C, et al., SGLT-1-mediated glucose uptake protects human intestinal epithelial cells against Giardia duodenalis-induced apoptosis, International journal for parasitology 38(8-9):923-934 (2008)).
  • SGLT1 and EGFR can synergistically promote prostate cancer growth.
  • prostate cancer cell lines e.g., PC3 and LNCaP (both positive for EGFR and SGLT1)
  • EGFR tyrosine kinase inhibitors e.g., Gefitnib, Erlotinib, Icontinib, Mubritinib, Vandertanib, Lapatinib, Pelitinib, Canertinib, Neratinib, Afatinib and Dacomitinib.
  • an SGLT1 inhibitor e.g., phlorizin, and phlorizin derivatives, such as Canagliflozin and Dapagliflozin
  • an SGLT1 inhibitor e.g., phlorizin, and phlorizin derivatives, such as Canagliflozin and Dapagliflozin
  • the growth inhibitory effects of the treatments may be determined.
  • cells can be treated with the SGLT1 inhibitor phlorizin (50 ⁇ M) with/without EGFR inhibitors (Gefitinib, 10 ⁇ M; Erlotinib, 10 ⁇ M) for 48 hs before being subjected to MTT assay.
  • the OD value of control cells can be artificially set as 1. All experiments may be repeated for at least 3 times.
  • Asterisk marks indicate statistical significances between linked groups.
  • FIG. 4B illustrates a Western blot analysis of the expressions of endogenous EGFR and SGLT1 in PC3 and LNCaP cells.
  • breast and colon cancer cells express EGFR and SGLT1.
  • FIG. 4C shows an MTT assay of the effect of inhibition of SGLT1 on the growth inhibitory effect of Gefitinib and Erlotinib on PC3 cells.
  • FIG. 4D shows an MTT assay of the effect of inhibition of SGLT1 on the growth inhibitory effect of Gefitinib and Erlotinib on LNCaP cells. Based on these results, in one embodiment, co-inhibition of SGLT1 and EGFR functions can more effectively inhibit cancer cell growth.
  • Phlorizin can significantly sensitize prostate cancer cells to the growth inhibitory effects of Gefitnib and Erlotinib.
  • Phlorizin or Phlorizin-like compounds can be administered to a patient, intravenously, intratumorally, or systemically, to treat cancer.
  • Phlorizin or Phlorizin-like compounds can be administered to a patient in conjunction with tyrosine-kinase inhibitors to treat cancer.
  • a peptide can destabilize EGFR and SGLT1 proteins of cancer cells.
  • FIG. 5A illustrates an embodiment of a peptide, called ESD-01 (SEQID: 001), which can destabilize EGFR and SGLT1 proteins of cancer cells.
  • the ESD-01 peptide can be selected from the SGLT1 interacting domain of EGFR.
  • ESD-01 comprises amino acids 1049-1062 of a human EGFR protein (GenBank: AAH94761.1).
  • the effects of ESD-01 on the stability of EGFR and SGLT1 in cultured cells can be determined by Western blot analysis.
  • FIG. 5B shows that ESD-01 treatment (20004 for 6 hours) significantly down-regulates EGFR and SGLT1 levels in PC3 cells, which are inhibited by the proteasome inhibitor MG132.
  • inhibition of SGLT1 and EGFR down-regulation by ESD-01 by MG132 suggests that MTG-01 destabilizes EGFR and SGLT1 via proteolysis.
  • the effects of ESD-01 on the survivability of cultured cancer cells can be determined by trypan blue uptake assay.
  • FIG. 6 shows that ESD-01 treatment (10004 for 24 hours) significantly reduces the survivability of a variety of cancer cells including prostate cancer PC3 and Du145 cells, breast cancer MDA-MB-231 cells, and colon cancer HCT116 cells.
  • ESD-01 treatment cannot reduce the survivability of non-cancerous HEK293 cells.
  • ESD-01 SEQID: 001
  • the amino acids can be either L-form or D-form chiral isomers.
  • Serine (S) in ESD-01 can be substituted with Threonine (T), Tyrosine (Y), or any other non-natural hydroxyl containing amino acid (e.g., SEQID: 002).
  • Threonine (T) in ESD-01 can be substituted with Serine (S), Tyrosine (Y), or any other non-natural hydroxyl containing amino acid (e.g., SEQID: 003).
  • Lysine (K) in ESD-01 can be substituted with Theronine (T) (e.g., SEQID: 004).
  • Glutamine (Q) can in ESD-01 can be substituted with Histidine (H) (e.g., SEQID: 005).
  • Threonine (T) at position 10 of ESD-01 can be substituted with Alanine (A) (e.g., SEQID: 006).
  • Serine (S) at position 12 of ESD-01 can be substituted with Leucine (L) (e.g., SEQID: 007).
  • L Leucine
  • the polar positive charged amino acids in ESD-01, Lysine (K) and Histidine (H) can be any other natural and non-natural positively charged amino acid (e.g., SEQID: 008).
  • the polar amino acids in ESD-01, Glutamine (Q) and Cysteine (C) can be any other natural and non-natural polar amino acid (e.g., SEQID: 009).
  • Cysteine (C) in ESD-01 can be any other natural and non-natural thiol side chain (—SH) containing amino acid (e.g., SEQID: 010).
  • the nonpolar amino acids in ESD-01, Leucine (L), Valine (V), and Tryptophan (W), can be any other natural and non-natural nonpolar amino acids (e.g., SEQID: 011).
  • HEK293 cell line, prostate cancer cell line PC3, LNCaP, Du145, MDA-MB-231, and HCT116 cells were originally obtained from the American Type of Culture Collection (ATCC) and maintained in DMEM supplemented with 10% fetal bovine serum and 1% Penicillin/Streptomycin under 5% CO 2 at 37° C.
  • Mouse anti-Flag-tag antibody (F1804), proteasome inhibitor MG231, and phlorizin dihydrate were obtained from Sigma-Aldrich (St. Louis, Mo.).
  • AEE788, Gefitinib, and Erlotinib were obtained from Selleckchem (Houston, Tex.).
  • Antibody against pEGFR (Y1173) (cat. no.
  • SGLT1-IHC immunohistochemical analysis
  • SGLT1-WB Western blotting analysis
  • Human wild type EGFR was cloned into a pcDNA3.1 vector (Clontech, CA), which was used as a parental vector to generate all the other EGFR constructs.
  • the pRK5 expression plasmid (Clontech, CA) with a c-terminal HA tag was used for the constructions of all HA tagged EGFRs.
  • the full-length human EGFR was amplified with a forward primer EGFR-F (ATTCTCGAGCGGGGAGCAGCGATG) and a reverse primer EGFR-R (CCTAAGCTTTGCTCCAATAAATTCACTG).
  • Primers for cloning the EGFR with extracellular domain deletion are ⁇ Extra-F: ATTCTCGAGATGTCC ATCGCCACTGGGATG and ⁇ Extra-R: CCTAAGCTTTGCTCCAATAAATTCACTGC; primers for intracellular deletion (Alntro, 671-1210 aa) are ⁇ Intra-F: TATCTCGAGATGCGACCCTCCGGGACGGC and ⁇ Intra-R: CCTAAGCTTCC TTCGCATGAAGAGGCC; primers for autophosphorylation domain deletion ( ⁇ Autophos, 978-1210aa) are ⁇ Autophospho-F: ATTCTCGAGATGTCCATCGCCACTGGGATG and ⁇ Autophospho-R: CCTAAGCTTGTAGCGCTGGGGGTCTCGG; primers for intracellular domain deletion
  • the kinase dead mutant of EGFR (KD-EGFR, R817M), transmembrane domain deletion (ATM, 645-670aa), and tyrosine kinase domain deletion (ATK, 670-977aa) plasmids were constructed from pRK5-WT-EGFR-HA by site-directed mutagenesis using the QuikChange Lightning Site-Directed Mutagenesis Kit (Agilent, CA) according to the manufacturer's protocol.
  • the primers were: KD-EGFR-F: GCACCGCGACCTGGCAGCC ATGAACGTACTGGTGAAAACACC and KD-EGFR-R: GGTGTTTTCACCAGTACGTTCATGGCTGCCA GGTCGCGGTGC; ⁇ TM-F: CGAGACCCCCAGCGCTACCGGACTCCCCTCCTGAGC and ⁇ TM-R: CGAGACCCCCAGCGCTACCGGACTCCCCTCCTGAGC; ⁇ TK-F: CGCTGCGGAGGCTGCTGCAGTAC CTTGTCATTCAGGGGG and ⁇ TK-R: CCCCCTGAATGCAAGGTACTGCAGCAGCCTCCGCAGCG. All of the constructs yielded fusion proteins with a C-terminal HA tag. All plasmids were confirmed by sequencing.
  • HEK293 cells were transfected with plasmids expressing flagged SGLT1 alone or with indicated HA tagged EGFR constructs. After 24 hours of transfection, cells were washed in 1 ⁇ phosphate buffered solution and lysed with RIPA buffer (50 mM Tris-HCl, pH 8.0, with 150 mM sodium chloride, 1.0% Igepal CA-630 (NP-40), 0.5% sodium deoxycholate, and 0.1% sodium dodecyl sulfate), supplemented with a protease inhibitors cocktail for 6 hours on a shaker at 4° C. The cell lysates were then centrifuged for 2 minutes at 12000 ⁇ rpm.
  • RIPA buffer 50 mM Tris-HCl, pH 8.0, with 150 mM sodium chloride, 1.0% Igepal CA-630 (NP-40), 0.5% sodium deoxycholate, and 0.1% sodium dodecyl sulfate
  • HEK293 cells were transfected with SGLT1 and wild type EGFR. After 18h, cells were starved in serum free medium for 6h before EGF treatment (10 ng/ml), or EGF plus AEE788 (5 ⁇ M) for 30-60 min. Control cells were treated with an equal volume of vehicle dimethyl sulfoxide (DMSO). Cell lysates were then subjected to immunoprecipitation as described above.
  • DMSO vehicle dimethyl sulfoxide
  • tissues were incubated with a mixture of Alexa Fluor 488-conjugated donkey anti-rabbit IgG and Alexa Flour 594-conjugated donkey anti-mouse IgG dissolved in PBS containing 10% donkey serum for 30 minutes at room temperature. The stained samples were then washed three times (5 minutes per wash) with PBS at room temperature. Fluorescence images were captured and analyzed with a confocal microscope (Olympus). Cell nucleus was stained by 4′,6-diamidino-2-phenylindole (DAPI).
  • DAPI 4′,6-diamidino-2-phenylindole
  • MTT 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium Bromide
  • the Student's t test was used to assess the difference in growth of cells treated with EGFR inhibitors in the presence/absence of SGLT1 inhibitor. P values less than 0.05 were defined as having statistical significance.

Abstract

A compound can destabilize a binding interaction between an epidermal growth factor receptor (EGFR) and a sodium/glucose co-transporter 1 (SGLT 1). In one embodiment, the compound is a peptide derived from the interacting domain of EGFR. In another embodiment, the peptide is administered to a patient to treat cancer.

Description

    CROSS-REFERENCES TO RELATED APPLICATIONS
  • This application claims priority to U.S. provisional application No. 61/821,028 filed on May 8, 2013, which is herein incorporated by reference in its entirety.
  • GOVERNMENT SPONSORSHIP American Cancer Society (RSG-09-206-01) Department of Defense Prostate Cancer Research Program (W91ZSQ8334N607) BACKGROUND
  • Epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase that is over-active/over-expressed in the majority of cancers of epithelial origin (Hynes N E, et al., ERBB receptors and cancer: the complexity of targeted inhibitors, Nature Reviews Cancer, 5(5):341-354 (2005)). Inhibition of the tyrosine kinase activity of EGFR has been the principle strategy of EGFR based cancer therapies. However, targeting EGFR by small molecule inhibitors of receptor tyrosine kinase has not produced satisfactory therapeutic efficacy. The general response rates are between 10-20% across a variety of human malignancies (Weiss J., First line erlotinib for NSCLC patients not selected by EGFR mutation: keep carrying the TORCH or time to let the flame die? Transl. Lung Cancer Res., 1(3):219-223 (2012); Cohen S J, et al., Phase II and pharmacodynamic study of the farnesyltransferase inhibitor R115777 as initial therapy in patients with metastatic pancreatic adenocarcinoma, J. Clinical Oncology, 21(7):1301-1306 (2003); Dancey J E, et al., Targeting epidermal growth factor receptor—are we missing the mark?, Lancet 362(9377):62-64 (2003)). In other words, there is major population of cancer patients that do not respond to EGFR tyrosine kinase inhibitors. For example, although EGFR is over-expressed in more than 80% of late stage prostate cancers and negatively correlates with prognosis, prostate cancer is resistant to EGFR inhibitors (Herres E., et al., Expression of the epidermal growth factor receptor family in prostate carcinoma before and during androgen-independence, British J. Cancer, 90(2):449-454 (2004); Pu Y S, et al., Epidermal growth factor receptor inhibitor (PD168393) potentiates cytotoxic effects of paclitaxel against androgen-independent prostate cancer cells, Biochemical Pharmacology, 71(6):751-760 (2006); Sherwood E R, et al., Epidermal growth factor-related peptides and the epidermal growth factor receptor in normal and malignant prostate, World J. Urology, 13(5):290-296 (1995); Zellweger T., et al., Expression patterns of potential therapeutic targets in prostate cancer, International J. Cancer, 113(4):619-628 (2005); Canil C M, et al., Randomized phase II study of two doses of gefitinib in hormone-refractory prostate cancer: a trial of the National Cancer Institute of Canada-Clinical Trials Group, J. Clinical Oncology, 23(3):455-460 (2005); Gross M, et al., A phase II trial of docetaxel and erlotinib as first-line therapy for elderly patients with androgen-independent prostate cancer, BMC Cancer 7:142 (2007)).
  • Evidence indicates that EGFR possesses tyrosine kinase independent functions. For example, EGFR knockout animals die soon after birth (Threadgill D W, et al., Targeted disruption of mouse EGF receptor: effect of genetic background on mutant phenotype, Science, 269(5221):230-234 (1995). However, mice with severely compromised EGFR tyrosine kinase activity are completely viable and display only some epithelial defects (Luetteke N C, et al., The mouse waved-2 phenotype results from a point mutation in the EGF receptor tyrosine kinase, Genes & Development, 8(4):399-413 (1994)). As another example, both a wild type and a kinase-dead EGFR enhanced the survival of EGFR negative 32D hematopoietic cells (Ewald J A, et al., Ligand- and kinase activity-independent cell survival mediated by the epidermal growth factor receptor expressed in 32D cells, Experimental Cell Research 282(2):121-131 (2003). It has also been discovered that EGFR participates in the maintenance of basal intracellular glucose level of cancer cells by interacting with and stabilizing the sodium-glucose co-transporter 1 (SGLT1), independent of EGFR tyrosine kinase activity (Weihua Z, et al., Survival of cancer cells is maintained by EGFR independent of its kinase activity, Cancer Cell, 13(5):385-393 (2008)).
  • SGLT1 is an active glucose transporter that relies on extracellular sodium concentration to transport glucose into cells independent of glucose concentration (Wright E M, et al., Biology of human sodium glucose transporters, Physiological Reviews, 91(2):733-794 (2011). SGLT1 plays a critical role in glucose absorption and retention in the body (Castaneda-Sceppa C, et al., Sodium-dependent glucose transporter protein as a potential therapeutic target for improving glycemic control in diabetes, Nutrition Reviews, 69(12):720-729 (2011)). One of the hallmarks of cancer is that cancer cells exhibit altered energy metabolism, i.e. cancer cells consume a substantially higher amount of nutrients and energy substrates than their normal counterparts (Hanahan D, et al., Hallmarks of cancer: the next generation, Cell, 144(5):646-674 (2011). This enhanced energy consumption demands a high rate of nutrients uptake, which is achieved by over-expression of plasma membrane transporters (Ganapathy V, et al., Nutrient transporters in cancer: relevance to Warburg hypothesis and beyond, Pharmacology & Therapeutics 121(1):29-40 (2009). Studies have found that SGLT1 is over-expressed in various types of cancers including ovarian carcinoma, oral squamous cell carcinoma, colorectal cancer, pancreatic cancer, and prostate cancer (Lai B, et al., Overexpression of SGLT1 is correlated with tumor development and poor prognosis of ovarian carcinoma, Archives of Gynecology and Obstetrics, 285(5):1455-1461 (2012); Hanabata Y, et al., Coexpression of SGLT1 and EGFR is associated with tumor differentiation in oral squamous cell carcinoma, Odontology/the Society of the Nippon Dental University, 100(2):156-163 (2012); Guo G F, et al., Overexpression of SGLT1 and EGFR in colorectal cancer showing a correlation with the prognosis, Medical Oncology 28 Suppl 1:S197-203 (2011); Casneuf V F, et al., Expression of SGLT1, Bcl-2 and p53 in primary pancreatic cancer related to survival, Cancer Investigation 26(8):852-859 (2008); Blessing A, et al., Sodium/Glucose Co-transporter 1 Expression Increases in Human Diseased Prostate, J. Cancer Sci. Ther. 4(9):306-312 (2012). As an example, late stage prostate cancers express elevated levels of EGFR and uptake a high amount of glucose (Herres E., et al., Expression of the epidermal growth factor receptor family in prostate carcinoma before and during androgen-independence, British J. Cancer, 90(2):449-454 (2004); Pu Y S, et al., Epidermal growth factor receptor inhibitor (PD168393) potentiates cytotoxic effects of paclitaxel against androgen-independent prostate cancer cells, Biochemical Pharmacology, 71(6):751-760 (2006); Sherwood E R, et al., Epidermal growth factor-related peptides and the epidermal growth factor receptor in normal and malignant prostate, World J. Urology, 13(5):290-296 (1995); Lee S T, et al., PET in prostate and bladder tumors, Seminars in Nuclear Medicine 42(4):231-246 (2012); Oyama N, et al., The increased accumulation of [18F]fluorodeoxyglucose in untreated prostate cancer, Japanese J. Clinical Oncology, 29(12):623-629 (1999)). A better understanding of the functional relationship between EGFR and SGLT1 may lead to identification of novel therapeutic targets for cancer therapy.
  • Thus, there is need in the art for methods and compositions that can adequately treat cancer cells resistant to EGFR tyrosine kinase inhibitors.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The foregoing summary, as well as the following detailed description, will be better understood when read in conjunction with the appended drawings. For the purpose of illustration only, there is shown in the drawings certain embodiments. It's understood, however, that the inventive concepts disclosed herein are not limited to the precise arrangements and instrumentalities shown in the figures.
  • FIG. 1A illustrates a schematic diagram of different constructs of human EGFR, in accordance with embodiments. FIG. 1B illustrates an immunoprecipitation coupled Western blot analysis of interactions between mutated EGFRs and SGLT1, in accordance with embodiments.
  • FIG. 2A illustrates a Western blot analysis of expression levels of SGTL1 in HEK293 cells co-transfected with the WT-EGFR, the KD-EGFR and the ΔAutophos-EGFR, in accordance with embodiments. FIG. 2B illustrates a densitometric quantification of bands in the Western blot of FIG. 2A, in accordance with embodiments. FIG. 2C illustrates a Western blot analysis of the effect of a proteasome inhibitor MG132 on the down-regulation of SGLT1 by ΔAutophos-EGFR, in accordance with embodiments. FIG. 2D illustrates a densitometric quantification of bands in the Western blot of FIG. 2C, in accordance with embodiments.
  • FIG. 3A illustrates an immunoprecipitation coupled Western blot analysis of interactions between EGFR-HA and SGLT1-Flag in HEK293 cells treated with EGF or AEE788, in accordance with embodiments. FIG. 3B illustrates immunoprecipitation coupled Western blot analysis of interactions between endogenous EGFR and SGLT1 in PC3 cells treated with EGF or AEE788, in accordance with embodiments.
  • FIG. 4A illustrates co-localization of SGLT1 and EGFR in prostate cancer tissues from a prostate cancer tissue array, in accordance with embodiments. FIG. 4B illustrates a Western blot analysis of expressions of endogenous EGFR and SGLT1 in PC3 and LNCaP cells, in accordance with embodiments. FIG. 4C illustrates an MTT assay showing the effect of inhibition of SGLT1 on the growth inhibitory effect of Gefitinib and Erlotinib on PC3 cells, in accordance with embodiments. FIG. 4D illustrates an MTT assay showing the effect of inhibition of SGLT1 on the growth inhibitory effect of Gefitinib and Erlotinib on LNCaP cells, in accordance with embodiments.
  • FIG. 5A illustrates disruption of EGFR-SGLT1 interaction by small molecules of peptides to destabilize both proteins, in accordance with embodiments. FIG. 5B illustrates that treatment of PC3 cells with an EGFR-SGLT1 disrupting peptide (MTG-01) significantly down-regulates EGFR and SGLT1 proteins, in accordance with embodiments.
  • FIG. 6 illustrates the viability of cancerous PC3, Du145, HCT116, and MDA-MB-231 cells and treatment of non-cancerous HEK293 cells when treated with MTG-01, in accordance with embodiments.
  • SUMMARY
  • A summary of certain embodiments disclosed herein is set forth below. It's understood that this section is presented merely to provide the reader with a brief summary of certain embodiments and that these descriptions are not intended to limit this application's scope. Indeed, this disclosure may encompass a variety of embodiments that may not be set forth herein.
  • The present application discloses methods and compositions for treating cancer. In one embodiment, a compound destabilizes a binding interaction between an epidermal growth factor receptor (EGFR) and a sodium/glucose co-transporter 1 (SGLT 1). In another embodiment, the compound is a Phlorizin-like compound or a peptide derived from the interacting domain of EGFR.
  • In an embodiment, a compound that destabilizes the binding interaction between EGFR and SGLT 1 is administered to a subject intratumorally, intravenously, or systemically. The compound may be a Phlorizin-like compound or a peptide derived from the interacting domain of EGFR. In yet another embodiment, the compound may be administered in conjunction with a tyrosine-kinase inhibitor.
  • DETAILED DESCRIPTION
  • Before explaining at least one embodiment in detail, it should be understood that the inventive concepts set forth herein are not limited in their application to the construction details or component arrangements set forth in the following description or illustrated in the drawings. It should also be understood that the phraseology and terminology employed herein are merely for descriptive purposes and should not be considered limiting.
  • It should further be understood that any one of the described features may be used separately or in combination with other features. Other invented systems, methods, features, and advantages will be or become apparent to one with skill in the art upon examining the drawings and the detailed description herein. It's intended that all such additional systems, methods, features, and advantages be protected by the accompanying claims.
  • All references cited in this application are incorporated by reference in their entirety.
  • Over-expression of epidermal growth factor receptor (EGFR) is associated with poor prognosis in malignant tumors. Sodium/glucose co-transporter 1 (SGLT1) is an active glucose transporter that is over-expressed in cancers including prostate cancer. It has been found that EGFR interacts with and stabilizes SGLT1 in cancer cells.
  • As explained in extensive detail below, the following embodiments have been identified in this application:
      • the critical micro-domain of EGFR that is required for its sufficient interaction with and mutual stabilization with SGLT1;
      • the effects of activation/inactivation of EGFR on EGFR-SGLT1 interaction;
      • the measured expression of EGFR and SGLT1 in prostate cancer tissues and cell lines;
      • the effect of inhibition of SGLT1 on the sensitivity of prostate cancer cells to EGFR tyrosine inhibitors;
      • the amino acid sequence of a synthesized peptide, ESD-01;
      • the effect of ESD-01 on the stability of EGFR and SGLT1 proteins in cancer cells; and
      • the effects of ESD-01 on survivability of non-cancer cells (HEK293) and several types of cancer cells cultured in vitro.
      • the ESD-01 peptide made of either L-amino acids or D-amino acids are equally effective in killing cancer cells in vitro.
  • In one embodiment, the autophosphorylation region (978-1210 amino acids) of EGFR is required for its sufficient interaction with SGLT1. This interaction is independent of EGFR's tyrosine kinase activity. Most importantly, in another embodiment, the EGFR-SGLT1 interaction is irresponsive to EGFR tyrosine kinase modulators (EGF and tyrosine kinase inhibitors). In yet another embodiment, EGFR and SGLT1 co-localize in prostate cancer tissues. In still another embodiment, inhibition of SGLT1 by a SGLT1 inhibitor (Phlorizin) sensitizes prostate cancer cells (PC3 and LNCaP) to EGFR inhibitors (Gefitnib and Erlotinib). In a further embodiment, ESD-01 (SEQID: 001) destabilizes the EGFR-SGLT1 interaction. All of this data suggests that EGFR in cancer cells can exit in two types of statuses—a tyrosine kinase modulator responsive status and an irresponsive status. Therefore, in an embodiment, SGLT1 is a protein involved in EGFR's functions that are irresponsive to EGFR tyrosine kinase inhibitors, and the EGFR-SGLT1 interaction may be a novel target for prostate cancer therapy.
  • EGFR Protein Region Required for Interaction with SGLT1
  • In one embodiment, to determine the EGFR protein region required for its interaction with SGLT1, a flag tagged SGLT123 and HA tagged EGFRs having a variety of mutations may be created (Blessing A, et al., Sodium/Glucose Co-transporter 1 Expression Increases in Human Diseased Prostate, J. Cancer Sci. Ther. 4(9):306-312 (2012)). FIG. 1A, by way of example only, illustrates a schematic diagram of human EGFR constructs that may be used to determine the EGFR protein region required for its interaction with SGLT1. For example, in some embodiments, the constructs may include: wild type EGFR (“WT”); kinase dead EGFR (R817M) (“KD”) (SEQID: 012); transmembrane domain deletion (645-670aa) (“ATM”) (SEQID: 013); extracellular domain deletion (1-644aa) (“ΔExtra”) (SEQID: 014); intracellular domain deletion (671-1210aa) (“ΔIntra”) (SEQID: 015); tyrosine kinase domain deletion (670-977aa) (“ΔTK”) (SEQID: 016); or autophosphorylation domain deletion (978-1210aa) (“ΔAutophos”) (SEQID: 017).
  • In an embodiment, the flagged SGLT1 and above identified HA tagged EGFRs can be transiently co-transfected into HEK293 cells. In another embodiment, SGLT1 can be immunoprecipitated using anti-flag antibodies. In yet another embodiment, Western blot analyses may be performed for HA tagged EGFRs. FIG. 1B, by way of example only, illustrates a Western blot analysis of interactions between mutated EGFRs and SGLT1, where IP=immunoprecipitation, IB=Immunoblot, and Input=expression levels of indicated exogenous proteins in HEK293 whole cell lysates used for the immunoprecipitation. As shown in FIG. 1B, deletion of the entire intracellular domain or the autophosphorylation domain of EGFR substantially diminishes its interaction with SGLT1. In one embodiment, the autophosphorylation domain of EGFR is required for its sufficient interaction with SGLT1.
  • Previously, using EGFR's extracellular domain and an intracellular domain that does not contain the EGFR TM domain, it was discoverred that the extracellular domain of EGFR interacted with SGLT1 better than its intracellular domain (Weihua Z, et al., Survival of cancer cells is maintained by EGFR independent of its kinase activity, Cancer Cell 13(5):385-393 (2008)). In an embodiment, to further characterize the EGFR-SGLT1 interaction at the plasma membrane, the TM domain may be included into the constructs of truncated EGFRs. In one embodiment, the TM containing intracellular domain of EGFR, especially the autophosphorylation domain, interacts more strongly with SGLT1 than its extracelluar domain. The discrepancy between this embodiment and the data shown in the previous report is very likely due to the lack of TM domain in the intracellular domain construct used in the previous study.
  • EGFR's Autophosphorylation Domain Required to Prevent Proteasome-Mediated SGLT1 Degradation
  • In an embodiment, to determine whether the autophosphorylation domain of EGFR is required to sustain the stability of SGLT1, the expression level of SGLT1 co-transfected with the WT-EGFR, the KD-EGFR, and the ΔAutoPhos-EGFR into HEK293 cells may be measured. For example, FIG. 2A, by way of example only, illustrates a Western blot analysis of expression levels of SGTL1 in HEK293 cells co-transfected with the WT-EGFR, the KD-EGFR and the ΔAutophos-EGFR. The same amounts of DNA plasmids of SGLT1 and EGFRs may be used in each group of treatments. Furthermore, control cells can be transfected with the same amount of DNA of the empty vector. Actin may be used as loading control. FIG. 2B, by way of example only, illustrates a densitometric quantification of bands in the Western blot of FIG. 2A. Asterisk marks indicate statistical significance between the linked representative groups from triplicate experiments.
  • As shown in FIGS. 2A-2B, in one embodiment, the level of SGLT1 in the WT-EGFR and the KD-EGFR transfected cells is much higher than that in the control vector or ΔAutoPhos-EGFR transfected cells. In another embodiment, the autophosphorylation domain of EGFR can maintain the expression level of SGLT1. In still another embodiment, the level of SGLT1 in the ΔAutoPhos-EGFR transfected cells is significantly lower than that of the control cells. In yet another embodiment, the loss of SGLT1 interaction with EGFR can promote down-regulation of SGLT1.
  • In an embodiment, to determine whether proteasome is involved in loss of interaction with EGFR induced down-regulation of SGLT1, SGLT1 and ΔAutoPhos-EGFR co-transfected HEK293 cells can be treated with a proteasome inhibitor, such as but not limited to MG231. For example, FIG. 2C illustrates a Western blot analysis of the effect of a proteasome inhibitor MG132 on the down-regulation of SGLT1 by ΔAutophos-EGFR. Actin may be used as a loading control. FIG. 2D, by way of example only, illustrates a densitometric quantification of bands in the Western blot of FIG. 2C. Asterisk marks indicate statistical significance between the linked representative groups from triplicate experiments.
  • As shown in FIGS. 2C-2D, by way of example only, in one embodiment MG231 can inhibit the down-regulation of SGLT1 in ΔAutoPhos-EGFR transfected cells. In another embodiment, the proteasome machinery is involved in the loss of interaction with EGFR induced SGLT1 down-regulation. In other words, proteasomes may cause the ultimate degradation of SGLT1 when the EGFR-SLGT1 interaction is compromised. In yet another embodiment, the proteasome machinery may be a potential therapeutic target alone or in combination with the EGFR-SLGT1 interaction.
  • In one embodiment, deleting the SGLT1 interacting domain in EGFR promotes the down-regulation of SGTL1 via the proteasome machinery. In another embodiment, this disruption of the EGFR-SGLT1 interaction in EGFR positive cancer cells can lead to down-regulation of SGLT1. Furthermore, previous data shows that knocking down SGLT1 by shRNA results in autophagic cell death of prostate cancer cells (Weihua Z, et al., Survival of cancer cells is maintained by EGFR independent of its kinase activity, Cancer Cell 13(5):385-393 (2008). In an embodiment, the EGFR-SGLT1 interaction can be a significant target to improve EGFR based therapy for cancer.
  • EGFR-SGLT1 Interaction is Irresponsive to EGFR Tyrosine Kinase Inhibitors
  • In an embodiment, the effects of EGFR tyrosine kinase inhibitors on EGFR's interaction with SGLT1 can be determined. In an embodiment, WT-EGFR and SGLT1 co-transfected HEK293 cells can be treated with either EGF or an EGFR tyrosine kinase inhibitor, AEE788. SGLT1 may be immunoprecipitated, and the levels of EGFR that were co-immunoprecipitated with SGLT1 can be measured. For example, FIG. 3A, by way of example only, illustrates an immunoprecipitation coupled Western blot analysis of interactions between EGFR-HA and SGLT1-Flag in HEK293 cells treated with EGF or AEE788, where EGFR=total EGFR, pEGFR=phosphorylated EGFR, IP=immunoprecipitation, IB=immunoblot, and Input=expression levels of indicated exogenous proteins in HEK293 whole cell lysates used for the immunoprecipitation. As shown in FIG. 3A, in one embodiment, neither EGF nor AEE788 significantly affects the EGFR-SGLT1 interaction. In yet another embodiment, the EGFR that co-precipitates with SGLT1 is not phosphorylated.
  • In one embodiment, to determine the effects of EGF and AEE788 on endogenous EGFR-SGLT1 interaction and the phosphorylation status of endogenous EGFR that interacts with SGLT1, endogenous SGLT1 of PC3 cells treated with EGF or AEE788 may be immunoprecipitated. In another embodiment, the phosphorylation status of the EGFR co-precipitated with SGLT1 can be measured. By way of example only, FIG. 3B illustrates an immunoprecipitation coupled Western blot analysis of interactions between endogenous EGFR and SGLT1 in PC3 cells treated with EGF or AEE788. In an embodiment, neither EGF nor AEE788 affect the EGFR-SGLT1 interaction. In yet another embodiment, the endogenous SGLT1 interacting EGFR is not phosphorylated. In still another embodiment, the EGFR-SGLT1 interaction is irresponsive to modulators of EGFR's tyrosine kinase activity. In yet a further embodiment, EGFR can be targeted for cancer therapy at its non-kinase functionality, including but not limited to its interaction with SGLT1.
  • It has been well documented that, upon phosphorylation of tyrosines within the autophosphorylation domain of EGFR, the autophosphorylation domain serves as a major docking site for recruitment of adaptor/effector proteins that transactivate downstream signaling (Bazley L A, et al., The epidermal growth factor receptor family, Endocrine-Related Cancer, 12 Suppl 1:S17-27 (2005)). Based on the results of FIG. 3A, in an embodiment, the EGFR-SGLT1 interaction is independent of EGFR activation/inactivation. The autophosphorylation domain of EGFR functions as a protein-protein interacting domain independent of EGFR's tyrosine kinase activity. In one embodiment, EGFR possesses pro-survival functions independent of its tyrosine kinase activity. For example, EGFR can exist in two types of statuses—a tyrosine kinase responsive status and a tyrosine kinase irresponsive status. Upon activation by EGFR's ligands, the autophosphorylation domain of the kinase responsive EGFR can be phosphorylated and can recruit effectors to trigger downstream signals. Alternatively, the kinase irresponsive EGFR may constantly interact with proteins regardless of the presence of EGFR ligands, and regardless of activation or inactivation of its tyrosine kinase. In yet another embodiment, SGLT1 may be one of such proteins that bind to and keep EGFRs in their kinase irresponsive status. In yet another embodiment, the non-phosphorylated autophosphorylation domain of EGFR may be used as a tool for identifying therapeutic targets.
  • Inhibition of SGLT1 by SGLT1 Inhibitor Sensitizes Prostate Cancer Cells to EGFR Inhibitors
  • In an embodiment, to determine the clinical relevance of the EGFR-SGLT1 interaction, immunofluorescent co-staining of EGFR and SGLT1 on a tissue microarray of prostate cancers (n=44) may be performed. For example, FIG. 4A, by way of example only, illustrates the results of three representative prostate cancer tissues from a prostate cancer tissue array. In an embodiment, SGLT1 (green) and EGFR (red) may be co-localized in the prostate tissue (e.g., orange or yellow, as indicated by arrows). In another embodiment, stromal cells may be positive for SGLT1, but negative for EGFR (e.g., indicated by arrow heads). In yet another embodiment, in EGFR positive cancer samples (n=41), SGLT1 co-localizes with EGFR in cancer cells, but not stromal cells.
  • In still another embodiment, the EGFR-SGLT1 interaction may contribute to the pathogenesis of prostate cancer. For example, in an embodiment, the co-localization of EGFR with SGLT1 in prostate cancer tissues can indicate that the EGFR-SGLT1 interaction is cancer relevant. Currently, at the clinic, EGFR tyrosine kinase inhibitors have not yet shown satisfactory therapeutic effects for prostate cancer (Canil C M, et al., Randomized phase II study of two doses of gefitinib in hormone-refractory prostate cancer: a trial of the National Cancer Institute of Canada-Clinical Trials Group, J. Clinical Oncology, 23(3):455-460 (2005); Gross M, et al., A phase II trial of docetaxel and erlotinib as first-line therapy for elderly patients with androgen-independent prostate cancer, BMC Cancer 7:142 (2007)). In one embodiment, considering the fact that EGFR expression correlates with disease progression of prostate cancer and the clinical unresponsiveness of prostate cancers to EGFR tyrosine kinase inhibitors, EGFR may contribute to the disease progression of prostate cancer independent of its tyrosine kinase activity. Additionally, previous findings conclude that (1) prostate cancer tissues have increased expression of SGLT1,23 (2) a loss of EGFR protein but not its tyrosine kinase activity sensitized prostate cancer cells to chemotherapeutic agent,29 and (3) a loss of EGFR induced autophagic cell death was mediated by down-regulation of SGLT1 protein (Blessing A, et al., Sodium/Glucose Co-transporter 1 Expression Increases in Human Diseased Prostate, J. Cancer Sci. Ther. 4(9):306-312 (2012); Xu S, et al., Loss of EGFR induced autophagy sensitizes hormone refractory prostate cancer cells to adriamycin, The Prostate. 17:1216-1224 (2011); Weihua Z, et al., Survival of cancer cells is maintained by EGFR independent of its kinase activity, Cancer Cell, 13(5):385-393 (2008)). In yet another embodiment, EGFR can promote prostate cancer progression via stabilizing SGLT1 to sustain the high demand of glucose of late stage cancer cells. This embodiment is not only supported by all the embodiments described herein, but is also supported by past data that over-expression of SGLT1 prevented renal epithelial cells and intestinal epithelial cells from apoptosis (Ikari A, et al., Sodium-dependent glucose transporter reduces peroxynitrite and cell injury caused by cisplatin in renal tubular epithelial cells, Biochimica et Biophysica Acta 1717(2):109-117 (2005); Yu L C, et al., SGLT-1-mediated glucose uptake protects human intestinal epithelial cells against Giardia duodenalis-induced apoptosis, International journal for parasitology 38(8-9):923-934 (2008)).
  • It is well known that an increase in glucose levels can activate EGFR, that SGLT1 is over-expressed in prostate cancer tissues, and that prostate cancer is resistant to EGFR inhibitors. (Han L, et al., High glucose promotes pancreatic cancer cell proliferation via the induction of EGF expression and transactivation of EGFR, PloS One 6(11):e27074 (2011); Blessing A, et al., Sodium/Glucose Co-transporter 1 Expression Increases in Human Diseased Prostate, J. Cancer Sci. Ther. 4(9):306-312 (2012); Canil C M, et al., Randomized phase II study of two doses of gefitinib in hormone-refractory prostate cancer: a trial of the National Cancer Institute of Canada-Clinical Trials Group, J. Clinical Oncology, 23(3):455-460 (2005); Gross M, et al., A phase II trial of docetaxel and erlotinib as first-line therapy for elderly patients with androgen-independent prostate cancer, BMC Cancer 7:142 (2007)). In an embodiment, SGLT1 and EGFR can synergistically promote prostate cancer growth. In one embodiment, to test whether inhibition of SGLT1 can sensitize prostate cancer cells to EGFR inhibitors, prostate cancer cell lines (e.g., PC3 and LNCaP (both positive for EGFR and SGLT1)), may be treated with EGFR tyrosine kinase inhibitors (e.g., Gefitnib, Erlotinib, Icontinib, Mubritinib, Vandertanib, Lapatinib, Pelitinib, Canertinib, Neratinib, Afatinib and Dacomitinib.) in the presence/absence of an SGLT1 inhibitor (e.g., phlorizin, and phlorizin derivatives, such as Canagliflozin and Dapagliflozin) (Ehrenkranz J R, et al., Phlorizin: a review, Diabetes/Metabolism Research and Reviews 21(1):31-38 (2005)). In one embodiment, the growth inhibitory effects of the treatments may be determined. For example, in an embodiment, cells can be treated with the SGLT1 inhibitor phlorizin (50 μM) with/without EGFR inhibitors (Gefitinib, 10 μM; Erlotinib, 10 μM) for 48 hs before being subjected to MTT assay. The OD value of control cells can be artificially set as 1. All experiments may be repeated for at least 3 times. Asterisk marks indicate statistical significances between linked groups.
  • FIG. 4B, by way of example only, illustrates a Western blot analysis of the expressions of endogenous EGFR and SGLT1 in PC3 and LNCaP cells. In one embodiment, breast and colon cancer cells express EGFR and SGLT1. FIG. 4C shows an MTT assay of the effect of inhibition of SGLT1 on the growth inhibitory effect of Gefitinib and Erlotinib on PC3 cells. FIG. 4D shows an MTT assay of the effect of inhibition of SGLT1 on the growth inhibitory effect of Gefitinib and Erlotinib on LNCaP cells. Based on these results, in one embodiment, co-inhibition of SGLT1 and EGFR functions can more effectively inhibit cancer cell growth. In still another embodiment, Phlorizin can significantly sensitize prostate cancer cells to the growth inhibitory effects of Gefitnib and Erlotinib. In an embodiment, Phlorizin or Phlorizin-like compounds can be administered to a patient, intravenously, intratumorally, or systemically, to treat cancer. In still another embodiment, Phlorizin or Phlorizin-like compounds can be administered to a patient in conjunction with tyrosine-kinase inhibitors to treat cancer.
  • In another embodiment, a peptide can destabilize EGFR and SGLT1 proteins of cancer cells. FIG. 5A, by way of example only, illustrates an embodiment of a peptide, called ESD-01 (SEQID: 001), which can destabilize EGFR and SGLT1 proteins of cancer cells. In one embodiment, the ESD-01 peptide can be selected from the SGLT1 interacting domain of EGFR. In another embodiment, ESD-01 comprises amino acids 1049-1062 of a human EGFR protein (GenBank: AAH94761.1). In still another embodiment, the effects of ESD-01 on the stability of EGFR and SGLT1 in cultured cells (e.g., PC3, MDA-MB-231, and HCT116 cells) can be determined by Western blot analysis. FIG. 5B, by way of example only, shows that ESD-01 treatment (20004 for 6 hours) significantly down-regulates EGFR and SGLT1 levels in PC3 cells, which are inhibited by the proteasome inhibitor MG132. In an embodiment, inhibition of SGLT1 and EGFR down-regulation by ESD-01 by MG132 suggests that MTG-01 destabilizes EGFR and SGLT1 via proteolysis. In still another embodiment, the effects of ESD-01 on the survivability of cultured cancer cells (e.g., PC3, MDA-MB-231, and HCT116 cells) can be determined by trypan blue uptake assay. FIG. 6, by way of example only, shows that ESD-01 treatment (10004 for 24 hours) significantly reduces the survivability of a variety of cancer cells including prostate cancer PC3 and Du145 cells, breast cancer MDA-MB-231 cells, and colon cancer HCT116 cells. In yet another embodiment, ESD-01 treatment cannot reduce the survivability of non-cancerous HEK293 cells.
  • It is well known that replacement of amino acids in a protein/peptide with different amino acids having similar chemical properties can generate proteins/peptides with identical functions. In an embodiment, ESD-01 (SEQID: 001) can be substituted with amino acids having similar chemical properties or any other mutations that will destabilize EGFR and SGLT1 proteins of cancer cells. For example, in one embodiment, the amino acids can be either L-form or D-form chiral isomers. In another embodiment, Serine (S) in ESD-01 can be substituted with Threonine (T), Tyrosine (Y), or any other non-natural hydroxyl containing amino acid (e.g., SEQID: 002). In yet another embodiment, Threonine (T) in ESD-01 can be substituted with Serine (S), Tyrosine (Y), or any other non-natural hydroxyl containing amino acid (e.g., SEQID: 003). In still another embodiment, Lysine (K) in ESD-01 can be substituted with Theronine (T) (e.g., SEQID: 004). In an embodiment, Glutamine (Q) can in ESD-01 can be substituted with Histidine (H) (e.g., SEQID: 005). In another embodiment, Threonine (T) at position 10 of ESD-01 can be substituted with Alanine (A) (e.g., SEQID: 006). In yet another embodiment, where the Serine (S) at position 12 of ESD-01 can be substituted with Leucine (L) (e.g., SEQID: 007). In still another embodiment, the polar positive charged amino acids in ESD-01, Lysine (K) and Histidine (H), can be any other natural and non-natural positively charged amino acid (e.g., SEQID: 008). In one embodiment, the polar amino acids in ESD-01, Glutamine (Q) and Cysteine (C), can be any other natural and non-natural polar amino acid (e.g., SEQID: 009). In another embodiment, the Cysteine (C) in ESD-01 can be any other natural and non-natural thiol side chain (—SH) containing amino acid (e.g., SEQID: 010). In still another embodiment, the nonpolar amino acids in ESD-01, Leucine (L), Valine (V), and Tryptophan (W), can be any other natural and non-natural nonpolar amino acids (e.g., SEQID: 011).
  • It's understood that the above description is intended to be illustrative, and not restrictive. The material has been presented to enable any person skilled in the art to make and use the inventive concepts described herein, and is provided in the context of particular embodiments, variations of which will be readily apparent to those skilled in the art (e.g., some of the disclosed embodiments may be used in combination with each other). Many other embodiments will be apparent to those of skill in the art upon reviewing the above description. The scope of the invention therefore should be determined with reference to the appended claims, along with the full scope of equivalents to which such claims are entitled. In the appended claims, the terms “including” and “in which” are used as the plain-English equivalents of the respective terms “comprising” and “wherein.”
  • Examples Cells and Reagents
  • HEK293 cell line, prostate cancer cell line PC3, LNCaP, Du145, MDA-MB-231, and HCT116 cells were originally obtained from the American Type of Culture Collection (ATCC) and maintained in DMEM supplemented with 10% fetal bovine serum and 1% Penicillin/Streptomycin under 5% CO2 at 37° C. Mouse anti-Flag-tag antibody (F1804), proteasome inhibitor MG231, and phlorizin dihydrate were obtained from Sigma-Aldrich (St. Louis, Mo.). AEE788, Gefitinib, and Erlotinib were obtained from Selleckchem (Houston, Tex.). Antibody against pEGFR (Y1173) (cat. no. 2434L) was obtained from Cell Signaling (Danvers, Mass.). Monoclonal antibody against C225 was obtained from Dr. Lee Elis (M.D. Anderson Cancer Center). Rabbit anti-actin (cat. no. sc-7210), rabbit anti-HA-tag antibody (sc-805), secondary antibodies against rabbit and mouse labeled with horseradish peroxidase, and protein A/G conjugated agarose beads (cat. no. sc-2003) were obtained from Santa Cruz Biotechnology (Santa Cruz, Calif.). MTT kit (cat. no. 30-1010K) was obtained from ATCC. The plasmid expressing flag tagged human SGLT1 and the rabbit anti-human-SGLT1 polyclonal antibodies for immunohistochemical analysis (SGLT1-IHC) and Western blotting analysis (SGLT1-WB) have been previously described (Blessing A, et al., Sodium/Glucose Co-transporter 1 Expression Increases in Human Diseased Prostate, J. Cancer Sci. Ther. 4(9):306-312 (2012)).
  • Plasmid Constructions.
  • Human wild type EGFR was cloned into a pcDNA3.1 vector (Clontech, CA), which was used as a parental vector to generate all the other EGFR constructs. The pRK5 expression plasmid (Clontech, CA) with a c-terminal HA tag was used for the constructions of all HA tagged EGFRs. The full-length human EGFR was amplified with a forward primer EGFR-F (ATTCTCGAGCGGGGAGCAGCGATG) and a reverse primer EGFR-R (CCTAAGCTTTGCTCCAATAAATTCACTG). DNA fragments were digested by Xho I and Hind III and cloned into the corresponding sites of the pRK5 vector. Primers for cloning the EGFR with extracellular domain deletion (ΔExtra, 1-644aa) are ΔExtra-F: ATTCTCGAGATGTCC ATCGCCACTGGGATG and ΔExtra-R: CCTAAGCTTTGCTCCAATAAATTCACTGC; primers for intracellular deletion (Alntro, 671-1210 aa) are ΔIntra-F: TATCTCGAGATGCGACCCTCCGGGACGGC and ΔIntra-R: CCTAAGCTTCC TTCGCATGAAGAGGCC; primers for autophosphorylation domain deletion (ΔAutophos, 978-1210aa) are ΔAutophospho-F: ATTCTCGAGATGTCCATCGCCACTGGGATG and ΔAutophospho-R: CCTAAGCTTGTAGCGCTGGGGGTCTCGG; primers for intracellular domain deletion (645-1210aa) are ΔIntra-F: TATCTCGAGATGCGACCCTCCGGGACGGC and ΔIntra-R: CCTAAGCTTCCTTCGCATGAAGAGGC. The kinase dead mutant of EGFR (KD-EGFR, R817M), transmembrane domain deletion (ATM, 645-670aa), and tyrosine kinase domain deletion (ATK, 670-977aa) plasmids were constructed from pRK5-WT-EGFR-HA by site-directed mutagenesis using the QuikChange Lightning Site-Directed Mutagenesis Kit (Agilent, CA) according to the manufacturer's protocol. The primers were: KD-EGFR-F: GCACCGCGACCTGGCAGCC ATGAACGTACTGGTGAAAACACC and KD-EGFR-R: GGTGTTTTCACCAGTACGTTCATGGCTGCCA GGTCGCGGTGC; ΔTM-F: CGAGACCCCCAGCGCTACCGGACTCCCCTCCTGAGC and ΔTM-R: CGAGACCCCCAGCGCTACCGGACTCCCCTCCTGAGC; ΔTK-F: CGCTGCGGAGGCTGCTGCAGTAC CTTGTCATTCAGGGGG and ΔTK-R: CCCCCTGAATGCAAGGTACTGCAGCAGCCTCCGCAGCG. All of the constructs yielded fusion proteins with a C-terminal HA tag. All plasmids were confirmed by sequencing.
  • Transient Transfection and Immunoprecipitation.
  • HEK293 cells were transfected with plasmids expressing flagged SGLT1 alone or with indicated HA tagged EGFR constructs. After 24 hours of transfection, cells were washed in 1× phosphate buffered solution and lysed with RIPA buffer (50 mM Tris-HCl, pH 8.0, with 150 mM sodium chloride, 1.0% Igepal CA-630 (NP-40), 0.5% sodium deoxycholate, and 0.1% sodium dodecyl sulfate), supplemented with a protease inhibitors cocktail for 6 hours on a shaker at 4° C. The cell lysates were then centrifuged for 2 minutes at 12000×rpm. Supernatant were then incubated with sepharose protein A/G beads conjugated with anti-flag or anti-SGLT1 antibody for overnight at 4° C. Samples were then centrifuged and washed with RIPA buffer three times before boiled in Laemmle buffer (Biorad, CA) and subjected to Western blot analysis. To determine the role of EGFR's tyrosine kinase in EGFR-SGLT1 interaction, HEK293 cells were transfected with SGLT1 and wild type EGFR. After 18h, cells were starved in serum free medium for 6h before EGF treatment (10 ng/ml), or EGF plus AEE788 (5 μM) for 30-60 min. Control cells were treated with an equal volume of vehicle dimethyl sulfoxide (DMSO). Cell lysates were then subjected to immunoprecipitation as described above.
  • Western Blot Analysis.
  • For Western blot (WB) analysis, cells were lysed with RIPA buffer (150 mM NaCl, 50 mM Tris-HCl, pH 7.4, 0.1% SDS, 1% TritonX-100, 1 mM EDTA, 1 mM PMSF, 20 μg/ml aprotinin, 20 μg/ml leupeptin, 20 μg/ml pepstatine, 1% sodium deoxycholate, 1 mM NaF, 1 mM Na3VO4, in H2O). Proteins separated by 8% SDS-PAGE were transferred to PVDF membrane followed by blocking with 5% nonfat dry milk and then incubation with primary antibodies at optimized concentrations for overnight at 4° C. The membranes were washed with 0.1% TBS/T (1×TBS, 0.1% Tween-20) 3 times, each time for 5 min before incubation with secondary antibody for 1 hour at room temperature. Signals were visualized by enhanced chemiluminescence.
  • Immunofluorescent Co-Staining.
  • For immunofluorescent co-staining of SGLT1 and EGFR, slides of prostate cancer tissue array were deparrafinated and rehydrated before antigens were retrieved in boiling citrate buffer for 10 min. Cooled tissue slides were then incubated in a blocking solution (5% donkey serum in PBS) for 1 hour at room temperature and then overnight at 4° C. with the rabbit polyclonal antibody against SGLT1 (SGLT1-IHC) (1:200 dilution) and C225 (1:200) in PBS containing 10% donkey serum (Blessing A, et al., Sodium/Glucose Co-transporter 1 Expression Increases in Human Diseased Prostate, J. Cancer Sci. Ther., 4(9):306-312 (2012)). After being washed three times with PBS, tissues were incubated with a mixture of Alexa Fluor 488-conjugated donkey anti-rabbit IgG and Alexa Flour 594-conjugated donkey anti-mouse IgG dissolved in PBS containing 10% donkey serum for 30 minutes at room temperature. The stained samples were then washed three times (5 minutes per wash) with PBS at room temperature. Fluorescence images were captured and analyzed with a confocal microscope (Olympus). Cell nucleus was stained by 4′,6-diamidino-2-phenylindole (DAPI).
  • Cell Growth Assay.
  • Cell growth was determined by 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium Bromide (MTT) assay in 96-well plates according to the protocol provided by the manufacture. Briefly, 5000 cells suspended in 100 μL medium were seeded in each well of a 96-well plate. On the second day, medium was replaced by medium containing Phlozidin (50 μM) and EGFR inhibitors (Iressa: 20 μM; Erlotinib: 20 μM). After 24 or 48 hours incubation with drugs, 10 μL MTT reagents was added to each well and incubated for 4 hours. After removal of the medium, the formazan precipitates in cells were dissolved in 100 μL DMSO. Absorbance was measured by a MultiSkan plate reader (Thermo Fisher Scientific, NC) at 570 nm. Triplicates of sample in each group were used. Cell viability was determined by trypan blue uptake assay. The percentage of live cells were counted in areas (n=3 for each sample) randomly selected under 10× magnificence of triplicates.
  • Statistical Analysis.
  • The Student's t test was used to assess the difference in growth of cells treated with EGFR inhibitors in the presence/absence of SGLT1 inhibitor. P values less than 0.05 were defined as having statistical significance.

Claims (20)

What is claimed is:
1. A composition comprising:
a compound that destabilizes a binding interaction between an epidermal growth factor receptor (EGFR) and a sodium/glucose co-transporter 1 (SGLT 1).
2. The composition of claim 1, wherein the compound is at least one of a peptide, and a peptide analog.
3. The composition of claim 2, wherein the peptide is derived from the SGLT 1 interacting domain of EGFR.
4. The composition of claim 2, wherein the peptide comprises the amino acid sequence LVWKQSCSSTSSTH (SEQID: 001).
5. The composition of claim 2, wherein the peptide comprises a compound substantially similar to at least one of SEQID: 002, SEQID: 003, SEQID: 004, SEQID: 005, SEQID: 006, SEQID: 007, SEQID: 008, SEQID: 009, SEQID: 0010, and SEQID: 0011.
6. The composition of claim 1, wherein the compound is at least one of a Phlorizin SGLT 1 inhibitor, and a Phlorizin-like SGLT 1 inhibitor.
7. A method of treating cancer cells in a subject comprising:
administering to the subject a compound that destabilizes a binding interaction between an epidermal growth factor receptor (EGFR) and a sodium/glucose co-transporter 1 (SGLT 1).
8. The method of claim 7, wherein the compound is at least one of a peptide, and a peptide analog.
9. The method of claim 8, wherein the peptide comprises the amino acid sequence LVWKQSCSSTSSTH (SEQID: 001).
10. The method of claim 8, wherein the peptide comprises at least one of SEQID: 002, SEQID: 003, SEQID: 004, SEQID: 005, SEQID: 006, SEQID: 007, SEQID: 008, SEQID: 009, SEQID: 0010, and SEQID: 0011.
11. The method of claim 7, further comprising administering to the subject a tyrosine-kinase inhibitor.
12. The method of claim 7, further comprising administering to the subject a compound that targets the proteasome machinery.
13. The method of claim 7, wherein the cancer cells are at least one of breast cancer cells, prostate cancer cells, and colon cancer cells.
14. The method of claim 8, further comprising identifying targets for cancer therapy by using the peptide to identify downstream survival pathways controlled by the EGFR-SGLT1 interaction.
15. A method of treating cancer cells in a subject comprising:
administering to the subject a tyrosine-kinase inhibitor and at least one of a Phlorizin SGLT 1 inhibitor, and a Phlorozin-like SGLT 1 inhibitor.
16. The method of claim 15, wherein the administering destabilizes a binding interaction between an epidermal growth factor receptor (EGFR) and a sodium/glucose co-transporter 1 (SGLT 1).
17. The method of claim 15, further comprising administering to the subject a tyrosine-kinase inhibitor.
18. The method of claim 17, wherein the cancer cells are prostate cancer cells.
19. The method of claim 17, wherein the tyrosine-kinase inhibitor is at least one of Gefitnib, Erlotinib, Icontinib, Mubritinib, Vandertanib, Lapatinib, Peletinib, Canetinib, Neratinib, Afatinib, and Dacomitinib.
20. The method of claim 15, wherein the Phlorizin SGLT 1 inhibitor is at least one of Canagliflozin, and Dapagliflozin.
US14/273,375 2013-05-08 2014-05-08 Targeting the EGFR-SGLT1 interaction for cancer therapy Active US9334307B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/273,375 US9334307B2 (en) 2013-05-08 2014-05-08 Targeting the EGFR-SGLT1 interaction for cancer therapy
US15/139,066 US9770482B2 (en) 2013-05-08 2016-04-26 Targeting the EGFR-SGLT1 interaction for cancer therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361821028P 2013-05-08 2013-05-08
US14/273,375 US9334307B2 (en) 2013-05-08 2014-05-08 Targeting the EGFR-SGLT1 interaction for cancer therapy

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/139,066 Division US9770482B2 (en) 2013-05-08 2016-04-26 Targeting the EGFR-SGLT1 interaction for cancer therapy

Publications (2)

Publication Number Publication Date
US20140336129A1 true US20140336129A1 (en) 2014-11-13
US9334307B2 US9334307B2 (en) 2016-05-10

Family

ID=51865227

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/273,375 Active US9334307B2 (en) 2013-05-08 2014-05-08 Targeting the EGFR-SGLT1 interaction for cancer therapy
US15/139,066 Active US9770482B2 (en) 2013-05-08 2016-04-26 Targeting the EGFR-SGLT1 interaction for cancer therapy

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/139,066 Active US9770482B2 (en) 2013-05-08 2016-04-26 Targeting the EGFR-SGLT1 interaction for cancer therapy

Country Status (5)

Country Link
US (2) US9334307B2 (en)
EP (1) EP2994155B1 (en)
JP (2) JP6309080B2 (en)
CN (2) CN115814056A (en)
WO (1) WO2014182955A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018026673A1 (en) * 2016-08-01 2018-02-08 IC-MedTech Corp. Ascorbic acid, quinone compound, and sodium glucose cotransporter inhibitor for treating cancer
CN113893256A (en) * 2020-07-06 2022-01-07 诺未科技(北京)有限公司 Application of compound or pharmaceutically acceptable salt, dimer or trimer thereof in preparation of medicine for treating cancer
EP3957329A4 (en) * 2020-07-06 2022-02-23 Newish Technology (Beijing) Co., Ltd. Composition for treating cancer, application thereof, and drug
EP3957310A4 (en) * 2020-07-06 2022-03-09 Newish Technology (Beijing) Co., Ltd. Composition and use thereof in preparation of medication for treating cancer

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016134486A1 (en) * 2015-02-27 2016-09-01 Steinberg Gregory Use of canagliflozin and derivatives thereof in the treatment of cancer
CN113905762A (en) * 2019-05-30 2022-01-07 北京生命科学研究所 Drug target of idiopathic pulmonary fibrosis
CN114569727B (en) * 2020-11-30 2023-12-01 诺未科技(北京)有限公司 Composition for treating cancer and application and medicine thereof
CN114569728B (en) * 2020-11-30 2023-11-28 诺未科技(北京)有限公司 Composition, application and medicine thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011091716A1 (en) * 2010-01-26 2011-08-04 上海市肿瘤研究所 Epidermal growth factor receptor variant

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4684627A (en) * 1981-09-08 1987-08-04 Leveen Harry H Treatment of cancer with phlorizin and its derivatives
CA1289077C (en) * 1984-08-13 1991-09-17 Harry H. Leveen Treatment of cancer with phlorizin and its derivatives
US5595756A (en) * 1993-12-22 1997-01-21 Inex Pharmaceuticals Corporation Liposomal compositions for enhanced retention of bioactive agents
DE10053496A1 (en) * 2000-10-27 2002-05-08 Peter Fuerst Compositions containing flavonoids and phloridzin or phloretin
US20100248249A1 (en) * 2007-08-17 2010-09-30 Allos Therapeutics, Inc. Methods for Assessing Cancer for Increased Sensitivity to 10-Propargyl-10-Deazaaminopterin by Assessing Egfr Levels
US20140271474A1 (en) * 2011-06-14 2014-09-18 The Regents Of The University Of California Inhibitor probes for imaging sodium-glucose cotransporters in health and disease
AR087701A1 (en) 2011-08-31 2014-04-09 Japan Tobacco Inc PIRAZOL DERIVATIVES WITH SGLT1 INHIBITING ACTIVITY

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011091716A1 (en) * 2010-01-26 2011-08-04 上海市肿瘤研究所 Epidermal growth factor receptor variant
US8703917B2 (en) * 2010-01-26 2014-04-22 Shanghai Cancer Institute Epidermal growth factor receptor variants and pharmaceutical compositions thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Zhang et al. (Survival of Cancer Cells in Maintained by EGFR Independent of Its Kinase Activity; Cancer Cell; 13, 385-393, May 2008). *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018026673A1 (en) * 2016-08-01 2018-02-08 IC-MedTech Corp. Ascorbic acid, quinone compound, and sodium glucose cotransporter inhibitor for treating cancer
CN113893256A (en) * 2020-07-06 2022-01-07 诺未科技(北京)有限公司 Application of compound or pharmaceutically acceptable salt, dimer or trimer thereof in preparation of medicine for treating cancer
EP3957329A4 (en) * 2020-07-06 2022-02-23 Newish Technology (Beijing) Co., Ltd. Composition for treating cancer, application thereof, and drug
EP3964217A4 (en) * 2020-07-06 2022-03-09 Newish Technology (Beijing) Co., Ltd. Use of compound or pharmaceutically acceptable salt, dimer or trimer thereof in preparation of drug for treating cancer
EP3957310A4 (en) * 2020-07-06 2022-03-09 Newish Technology (Beijing) Co., Ltd. Composition and use thereof in preparation of medication for treating cancer

Also Published As

Publication number Publication date
CN105358173A (en) 2016-02-24
US9770482B2 (en) 2017-09-26
WO2014182955A3 (en) 2015-10-29
EP2994155A2 (en) 2016-03-16
EP2994155B1 (en) 2020-12-23
JP6706635B2 (en) 2020-06-10
EP2994155A4 (en) 2017-03-29
JP2016520066A (en) 2016-07-11
CN115814056A (en) 2023-03-21
JP6309080B2 (en) 2018-04-18
JP2018052990A (en) 2018-04-05
US20160228495A1 (en) 2016-08-11
CN105358173B (en) 2022-09-02
US9334307B2 (en) 2016-05-10
WO2014182955A2 (en) 2014-11-13

Similar Documents

Publication Publication Date Title
US9770482B2 (en) Targeting the EGFR-SGLT1 interaction for cancer therapy
Zhang et al. OTUB2 promotes cancer metastasis via hippo-independent activation of YAP and TAZ
Tang et al. Selective inhibition of STRN3-containing PP2A phosphatase restores hippo tumor-suppressor activity in gastric cancer
Hua et al. TRB3 links insulin/IGF to tumour promotion by interacting with p62 and impeding autophagic/proteasomal degradations
Liang et al. Recombinant human erythropoietin antagonizes trastuzumab treatment of breast cancer cells via Jak2-mediated Src activation and PTEN inactivation
Seshacharyulu et al. Targeting EGF-receptor (s)-STAT1 axis attenuates tumor growth and metastasis through downregulation of MUC4 mucin in human pancreatic cancer
Jin et al. Desmoglein-2 modulates tumor progression and osimertinib drug resistance through the EGFR/Src/PAK1 pathway in lung adenocarcinoma
Nguyen et al. A selective inhibitor of ubiquitin-specific protease 4 suppresses colorectal cancer progression by regulating β-catenin signaling.
Tailor et al. Y box binding protein 1 inhibition as a targeted therapy for ovarian cancer
Lee et al. Tissue transglutaminase activates cancer-associated fibroblasts and contributes to gemcitabine resistance in pancreatic cancer
Liu et al. Niclosamide inhibits epithelial-mesenchymal transition and tumor growth in lapatinib-resistant human epidermal growth factor receptor 2-positive breast cancer
Luo et al. PHLDB2 mediates cetuximab resistance via interacting with EGFR in latent metastasis of colorectal cancer
Chen et al. ATP6V0D1 promotes alkaliptosis by blocking STAT3-mediated lysosomal pH homeostasis
Wang et al. Cancer‐derived IgG involved in cisplatin resistance through PTP‐BAS/Src/PDK1/AKT signaling pathway
Jia et al. CD166-mediated epidermal growth factor receptor phosphorylation promotes the growth of oral squamous cell carcinoma
Montero et al. PDCD4 limits prooncogenic neuregulin-ErbB signaling
Xiao et al. USP39-mediated deubiquitination of Cyclin B1 promotes tumor cell proliferation and glioma progression
US9303069B2 (en) Peptides for treating cancer
You et al. SDC2 Stabilization by USP14 Promotes Gastric Cancer Progression through Co-option of PDK1
Menendez et al. Heregulin-triggered Her-2/neu signaling enhances nuclear accumulation of p21WAF1/CIP1 and protects breast cancer cells from cisplatin-induced genotoxic damage
Liu et al. The deubiquitinating enzyme MINDY2 promotes pancreatic cancer proliferation and metastasis by stabilizing ACTN4 expression and activating the PI3K/AKT/mTOR signaling pathway
Zhao et al. Genome-wide identification of CDC34 that stabilizes EGFR and promotes lung carcinogenesis
Hsieh et al. Combination treatment of arsenic trioxide and osimertinib in recurrent and metastatic head and neck squamous cell carcinoma
Dong-Xiang et al. Cyclin-dependent kinase like 3 promotes triple-negative breast cancer progression via inhibiting the p53 signaling pathway.
McCormick A Comparative Study of Radiological Threat Environments and Radiation Control

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE UNIVERSITY OF HOUSTON SYSTEM, TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ZHANG, WEIHUA;REEL/FRAME:036662/0383

Effective date: 20150915

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 4

AS Assignment

Owner name: UNITED STATES GOVERNMENT, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF HOUSTON;REEL/FRAME:064067/0123

Effective date: 20230616

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2552); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 8